The effect of cimetidine on PBL from healthy donors and melanoma patients: Augmentation of T cell responses to TCGF mitogens and alloantigens and of TCGF production

1986 ◽  
Vol 21 (2) ◽  
Author(s):  
Avi Eisenthal ◽  
Jehudith Monselise ◽  
Rachel Zinger ◽  
Aliza Adler
2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A167-A167
Author(s):  
Divya Lenkala ◽  
Jessica Kohler ◽  
Brian McCarthy ◽  
Michael Nelson ◽  
Jonathan McGee ◽  
...  

BackgroundNeoantigens are tumor-specific antigens that are important in the anti-tumor immune response. These antigens are not subject to central immune tolerance and are therefore potentially more immunogenic than tumor-associated antigens. NEO-STIM®, our propriety ex vivo induction process, was developed to generate T-cell products specific to these neoantigens from the peripheral blood of patient. Here, we present the results of a proof of concept, pre-clinical study with multiple successful process engineering runs generating a neoantigen-specific T-cell product (NEO-PTC-01) using leukaphereses from metastatic melanoma patients. These products contain specific T-cell responses targeting multiple neoantigens from each individual patient‘s tumor.MethodsPatient-specific neoantigens were predicted using our RECON® bioinformatics platform. Predicted high-quality neoantigens were utilized in our ex vivo stimulation protocol, NEO-STIM, in the process engineering runs of NEO-PTC-01. NEO-STIM is used to prime, activate and expand memory and de novo T-cell responses from both the CD4+ and the CD8+ compartment. High throughput flow cytometric analysis was performed to characterize the specificity and functionality (cytokine production and cytolytic capacity) of the induced T-cell responses.ResultsHere we present the successful induction of 4–5 CD8+ and 4–7 CD4+ T-cell responses per patient, generated using peripheral blood mononuclear cells from multiple melanoma patients during these successful process engineering runs. We then extensively characterized these T-cell responses and demonstrate that these responses are functional, specific and have cytolytic capacity. Moreover, the induced T cells can recognize autologous tumor.ConclusionsNEO-STIM is a novel platform that generates ex vivo T-cell responses to high-quality neoantigen targets. NEO-PTC-01, the neoantigen-specific T cell product generated from this process, is a potent adoptive cell therapy targeting multiple immunogenic neoantigens in patients with metastatic melanoma.


2011 ◽  
Vol 61 (2) ◽  
pp. 169-179 ◽  
Author(s):  
Ray Wilkinson ◽  
Katherine Woods ◽  
Rachael D’Rozario ◽  
Rebecca Prue ◽  
Frank Vari ◽  
...  

2004 ◽  
Vol 2 (8) ◽  
pp. 81
Author(s):  
D. Speiser ◽  
D. Lienard ◽  
V. Rubio-Godoy ◽  
E. Devevre ◽  
A.M. Krieg ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5431-5431
Author(s):  
Stickel S. Juliane ◽  
Claudia Berlin ◽  
Daniel J. Kowalewski ◽  
Heiko Schuster ◽  
Lothar Kanz ◽  
...  

Abstract Data regarding the graft-versus-leukemia (GVL) effect after allogeneic stem cell transplantation (SCT) and donor lymphocyte infusion strongly suggest that T lymphocytes play a major role in the rejection of leukemic cells. Immunotherapy directed against leukemia- associated antigens might elicit specific immune responses that may serve to eliminate minimal residual disease after chemotherapy, or enhance the GVL effect after SCT. To achieve this goal there is need to identify appropriate leukemia associated HLA ligands, which are able to induce specific T cell responses. We here aimed to characterize the HLA class I ligandome in AML patients to provide novel tumor associated antigens (TAA) for peptide-based immunotherapy employing our recently implemented approach of direct isolation and identification of naturally presented HLA ligands by affinity chromatography and mass spectrometry (LC-MS/MS) in AML (Stickel et.al., abstract in Blood 2012). Absolute HLA surface expression on AML cells and autologous monocytes and granulocytes was quantified by flow cytometry. HLA class I ligands were isolated from AML cells as well as bone marrow and peripheral blood mononuclear cell (BMNCs/PBMCs) of healthy donors. LC-MS/MS peptide analysis provided qualitative and semi-quantitative information regarding the composition of the respective ligandomes. Comparative analysis of malignant and benign samples served to identify ligandome-derived TAA (LiTAA) and to select peptide vaccine candidates. The most abundantly detected peptide candidates were checked for immunogenicity by ELISpot and confirmed by intracellular interferon-g staining of CD8+ T-cells. Meanwhile 15 AML patients (8 FLT3-ITD mutant) and 35 healthy donors were analyzed. We observed overexpression of HLA class I and II on AML cells as compared to autologous monocytes and granulocytes, with the level of significance reached for HLA class II (p=0,04). A total of more than 12,000 AML derived HLA ligands representing >6,000 different source proteins were identified; of which 2,220 were exclusively represented in AML, but not in healthy PBMC/BMNC. Data mining for broadly represented LiTAA pinpointed 98 TAA as most promising targets. HLA ligands derived from these TAA were presented exclusively on more than 33% of all analyzed AML samples, amongst them already described TAA (e.g. JUP, FAF1) as well as several new leukemia-associated proteins (e.g. MTCH2, METTL7A). Subset analysis of the FLT3-ITD positive AML cohort revealed 21 LiTAA presented exclusively on more than 50% of FLT3-ITD positive AML cases. Additional screening for HLA ligands derived from described leukemia associated antigens revealed overrepresentation for e.g. FLT3, NUSAP, RHAMM and RGS5. Specific CD8+ T cell responses were detected against two A*03 epitope pools (pool 1: APLP2, DKGZ, FAF1, MTCH2; pool 2: KLF2, METTL7A, VCIP1, WIPI1) in AML patients. Notably, the chosen A*03 epitope pools did not elicit specific responses of CTL from healthy donors. Taken together, our HLA class I ligandome analysis in AML for the first time identified naturally presented HLA ligands from patients including a vast array of new leukemia associated antigens representing promising targets for a multipeptide-based immunotherapy approach in AML. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 101 (Supplement 2) ◽  
pp. 14631-14638 ◽  
Author(s):  
C. Lonchay ◽  
P. van der Bruggen ◽  
T. Connerotte ◽  
T. Hanagiri ◽  
P. Coulie ◽  
...  

2010 ◽  
Vol 33 (8) ◽  
pp. 848-858 ◽  
Author(s):  
Daniel E. Speiser ◽  
Katrin Schwarz ◽  
Petra Baumgaertner ◽  
Vania Manolova ◽  
Estelle Devevre ◽  
...  

2021 ◽  
Author(s):  
Xiaoxiao Jin ◽  
Ding Yan ◽  
Sun Shihui ◽  
Xinyi Wang ◽  
Zining Zhou ◽  
...  

AbstractWhile SARS-CoV-2-specific T cells have been characterized to play essential roles in host immune protection in COVID-19 patients, few researches focus on the functional validation of T cell epitopes and development of vaccines inducing specific T cell responses. In this study, 120 CD8+ T cell epitopes from E, M, N, S and RdRp proteins of SARS-CoV-2 were validated by on-silicon prediction, DC-peptide-PBL costimulation with healthy donors’ PBMCs and HLA-A molecule competitive binding experiments. Among them, 110, 15, 6, 14 and 12 epitopes were highly homologous with SARS-CoV, OC43, NL63, HKU1, and 229E, respectively. Thirty-one epitopes restricted by HLA-A2 molecule were used to generate peptide cocktail vaccines in combination with Poly(I:C), R848 or polylactic-co-glycolic acid nanoparticles, which elicited robust specific CD8+ T cell responses in wild-type and HLA-A2/DR1 transgenic mice. Seven of the 31 epitopes were found to be cross-presented by HLA-A2 and H-2K/Db molecules. These data have provided a library of SARS-CoV-2 CD8+ T cell epitopes which restricted by a series of high-frequency HLA-A allotypes and covered broad population in Asia, and initially confirmed the feasibility of human MHC class I molecule-restricted SARS-CoV2 epitope peptide cocktail vaccines, thus will facilitate the development of T cell epitope vaccines and specific cellular function detection kits.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1805-1805
Author(s):  
Gunnar Kvalheim ◽  
Steinar Aamdal ◽  
Gustav Gaudernack

Abstract Anti-tumour vaccines targeting the entire tumour antigen repertoire represent an attractive immunotherapeutic approach. This repertoire is present in the total mRNA isolated from the tumour. The mRNA from each cancer patient may be amplified and thus overcome the problem of limitation of material that has hampered the development of individualized vaccines. We have developed immuno-gene-therapy for malignant melanoma and prostate cancer. Monocyte-derived dendritic dells are transfected with autologous melanoma-mRNA or mRNA from three prostate cancer cell lines (DU-145, LN-CaP and PC-3). The vaccines may generate T cell responses against a broad repertoire of tumor-associated epitopes, and the melanoma approach moreover target patient-specific tumor antigens. Effective protocols were established for mRNA-transfection by square wave electroporation and for generation of clinical grade dendritic cells. A full scale preclinical evaluation demonstrated in vitro T cell responses in 6/6 advanced melanoma patients. The responses were specific to antigens encoded by the transfected tumor-mRNA. Recently, we have conducted two phase I/II trials, in advanced malignant melanoma and androgen-resistant prostate cancer. Successful vaccine preparations were obtained for all 41 patients elected. No serious adverse effects were observed. Specific T cell responses (T cell proliferation and/or IFNγ ELISPOT) were demonstrated in 9/19 evaluable melanoma patients and in 12/19 prostate cancer patients. The response rates were higher for patients receiving intradermal vaccination, compared to intranodal injection. Thirteen prostate cancer patients developed a decrease in log-slope PSA. The PSA-response was significantly related to the T cell response (p=0,002). We conclude that the tumour mRNA based DC-vaccine is feasible and safe, and that T cell responses are elicited in about 50% of patients. In the next generation clinical protocols, patients will undergo Treg depletion by chemotherapy (Temozolomide) prior to DC vaccination. Subsequently, T cells will be expanded ex vivo using the Dynabeads Clin Ex Vivo system before re-infusion into the patients.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 464-464
Author(s):  
Yishay Ofran ◽  
Vladimir Brusic ◽  
H. T Kim ◽  
Robert J. Soiffer ◽  
J. H Antin ◽  
...  

Abstract Minor histocompatibility antigens (mHA) are targets of graft versus host disease (GVHD) and graft versus leukemia (GVL) responses. In male patients with female donors, Y chromosome-encoded mHA are known to be highly immunogenic but few MHC class I presented epitopes have been identified and T cell responses to HY mHA have not been examined in large cohorts of patients. We recently developed a novel method for rapid identification of putative mHA based on in-silico prediction of potential MHC class I restricted peptides. Criteria for HY mHA prediction included the following: Y chromosome encoded gene sequence; high affinity binding to HLA A*0201 (≤100nM); lack of identical sequence in homologous X gene; validated protein expression not restricted to male-specific tissues; and amino acid disparities between Y and X chromosome homologues predicted to be immunogenic. In this study, 43 peptide epitopes (9–10 amino acids) representing 5 Y-encoded proteins (DBY, SMCY, UTY, PCDH11Y, USP9Y) were identified. The only previously known Y-encoded (SMCY) HLA A*0201 restricted mHA (FIDSYICQV) ranked first in the list of predicted epitopes. Two peptides failed synthesis and 41 HY peptides were tested for T cell reactivity in post-transplant samples from 21 male patients with female donors (M-F), 9 male patients with male donors (M-M) and 19 healthy donors (7 males; 12 females). All patients and donors were HLA A*0201+ and all patients had chronic GVHD. T cell reactivity was determined by ELISPOT. Peripheral blood mononuclear cells were stimulated with individual peptides for 7 days. IFN-g secreting T cells were enumerated after 18 hour restimulation with HLA A*0201 T2 cells pulsed with the same peptide. Known HLA A*0201 restricted peptides from EBV, CMV, HIV and HCV were used as positive and negative controls. T cell responses to individual peptides could be blocked by anti human HLA class I antibodies but not by anti HLA class II antibodies. Using the recursive partitioning method, a minimum of 20 spots/2×105 PBMC above background level was identified as an optimal cut-off point to classify positive responses for the majority of peptides. T cell responses to some HY peptides were also detected in M-M patients and healthy donors, but responses were most frequent and of greater magnitude in M-F patients. The median number of positive peptide responses per patient was 8 in the M-F patient group, 1 in M-M patients and 2 in healthy donors (p=0.002 for M-F vs M-M, p=0.02 for M-F vs healthy donors). All M-F patients responded to at least 1 HY peptide whereas 3/9 (33%) M-M patients and 7/19 (37%) healthy donors did not respond to any peptides. 18/21 (86%) M-F patients responded to 3 or more peptides but this level of reactivity was only detected in 2/9 (22%) M-M patients and 7/19 (37%) healthy donors. High frequency responses (>50 spots/2×105 PBMC) were detected in 19/21 (91%) M-F patients but only in 2/9 (22%) M-M patients, 1/7 (14%) healthy males and 4/12 (33%) healthy females. T cell responses were detected in at least 1 M-F patient for 36 of 41 peptides and responses were detected against all 5 Y-encoded proteins. However, a subset of 20 peptides appeared to be highly immunogenic with T cells responses detected in >25% of M-F patients. No single HY peptide elicited responses in all M-F patients including the previously known HY mHa, which was only positive in 40% of M-F patients. In fact 9 other peptides derived from DBY (1), SMCY (3), UTY (3), PCDH11Y (1) and USP9Y (1) elicited responses in 40–71% of M-F patients. Each of these 10 peptides elicited high frequency responses (>50 spots/2×105 PBMC) in at least 3 M-F patients but not in any M-M patients or healthy males. Within the M-F patients, the frequency of response was not associated with severity of cGVHD, underlying hematologic disease, age, stem cell source, transplant conditioning regimen or donor type. There was a correlation between time post transplant and the number of HY peptide responses (r=0.53, p=0.002 for all F-M and M-M patients combined). The functional application of bioinformatic models represents a new approach for identifying large numbers of novel HY peptides and assessing T cell responses after transplantation. These studies demonstrate a highly diverse T cell response despite identical mismatch and HLA type. Extending this method to other HLA alleles and to autosomal genetic disparities will improve our understanding of the role of mHA in GVL and GVHD after allogeneic transplantation.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A648-A648
Author(s):  
Kelly-Anne Masterman ◽  
Oscar Haigh ◽  
Kirsteen Tullett ◽  
Ingrid Leal-Rojas ◽  
Carina Walpole ◽  
...  

BackgroundDendritic cells (DC) are crucial for the efficacy of cancer vaccines, but current vaccines do not harness the key cDC1 subtype required for effective CD8+ T cell mediated tumor immune responses. Vaccine immunogenicity could be enhanced by specific delivery of immunogenic tumor antigens to CD141+ DC, the human cDC1 equivalent. CD141+ DC exclusively express the C-type-lectin-like receptor CLEC9A, which is important for the regulation of CD8+ T cell responses. This study developed a new vaccine that harnesses a human anti-CLEC9A antibody to specifically deliver the immunogenic tumor antigen, NY-ESO-1 to human CD141+ DC. The ability of the CLEC9A-NY-ESO-1 antibody to activate NY-ESO-1 specific naïve and memory CD8+ T cells was examined and compared to a vaccine comprised of a human DEC-205-NY-ESO-1 antibody that targets all human DC.MethodsHuman anti-CLEC9A, anti-DEC-205 and isotype control IgG4 antibodies were genetically fused to NY-ESO-1 polypeptide. Cross-presentation to NY-ESO-1- epitope specific CD8+ T cells and reactivity of T cell responses in melanoma patients was assessed by IFNγ production following incubation of CD141+ DC and patient peripheral blood mononuclear cells with targeting antibodies. Humanized mice containing human DC subsets and a repertoire of naïve NY-ESO-1-specific CD8+ T cells were used to investigate naïve T cell priming. T cell effector function was measured by expression of IFNγ, MIP-1β, TNF and CD107a and by lysis of target tumor cells.ResultsCLEC9A-NY-ESO-1 Ab were effective at mediating delivery and cross-presentation of multiple NY-ESO-1 epitopes by CD141+ DC for activation of NY-ESO-1-specific CD8+ T cells. When benchmarked to NY-ESO-1 conjugated to an untargeted control antibody or to anti-human DEC-205, CLEC9A-NY-ESO-1 was superior at ex vivo reactivation of NY-ESO-1-specific T cell responses in melanoma patients. Moreover, CLEC9A-NY-ESO-1 induced priming of naïve NY-ESO-1-specific CD8+ T cells with polyclonal effector function and potent tumor killing capacity in vitro.ConclusionsThese data advocate human CLEC9A-NY-ESO-1 antibody as an attractive strategy for specific targeting of CD141+ DC to enhance tumour immunogenicity in NY-ESO-1-expressing malignancies.Ethics ApprovalWritten informed consent was obtained for human sample acquisition in line with standards established by the Declaration of Helsinki. Study approval was granted by the Mater Human Research Ethics Committee (HREC13/MHS/83 and HREC13/MHS/86) and The U.S. Army Medical Research and Materiel Command (USAMRMC) Office of Research Protections, Human Research Protection Office (HRPO; A-18738.1, A-18738.2, A-18738.3). All animal experiments were approved by the University of Queensland Animal Ethics Committee and conducted in accordance with the Australian Code for the Care and Use of Animals for Scientific Purposes in addition to the laws of the United States and regulations of the Department of Agriculture.


Sign in / Sign up

Export Citation Format

Share Document